1
|
Abdallah MA, Ibrahem EO, Ali EA, Tayel SG, Abo-Elsoud RAA. The antihypertensive effect of the coadministration of exercise and eugenol in deoxycorticosterone acetate-induced hypertension in rats. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04119-6. [PMID: 40261349 DOI: 10.1007/s00210-025-04119-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 03/28/2025] [Indexed: 04/24/2025]
Abstract
Hypertension (HTN) is a significant health issue with high morbidity, especially in the cardiovascular system, leading to mortality. HTN could be treated with exercise (Ex) and eugenol due to their antioxidant, anti-inflammatory, and antiapoptotic activities. 48 male Wistar rats (n = 8) were assigned to normal, deoxycorticosterone acetate (DOCA), losartan + DOCA, Ex + DOCA, eugenol + DOCA, and Ex + eugenol + DOCA groups. Blood pressure (BP), echocardiography, serum electrolytes, lipid profile, total antioxidant capacity (TAC), interleukin- 6 (IL- 6), endocan, endothelin- 1 (ET- 1), transforming growth factor-β1 (TGF-β1), plasma renin activity (PRA), and serum aldosterone were measured. PCR analyzed p38 mitogen-activated protein kinase (p38MAPK), c-Jun N-terminal kinases (JNK), and collagen 1 expression. Histopathology was used to evaluate heart and kidney tissues. Cardiac apoptosis was assessed using TUNEL staining. Losartan, aerobic Ex (30 min/day, 5 days/week), combined eugenol and Ex, and eugenol therapy mitigated systolic blood pressure (SBP), diastolic blood pressure (DBP), mean arterial pressure (MAP), heart rate (HR), left ventricular end-diastolic volume (LVEDV), serum sodium, IL- 6, endocan, ET- 1, TGF-β1, PRA, and serum aldosterone. These therapies attenuated histopathological changes, cardiac apoptosis, p38MAPK, JNK, and collagen 1. Conversely, ejection fraction (EF), serum potassium, and TAC levels were elevated compared to the DOCA group. Ex and eugenol monotherapy ameliorated DOCA-induced HTN in rats. Ex + eugenol combination showed equal efficacy to losartan in ameliorating DOCA-induced HTN, with better efficacy than Ex or eugenol alone. The combination's antihypertensive effect may result from the synergistic/additive antioxidant, anti-inflammatory, and antiapoptotic effects of either therapy.
Collapse
Affiliation(s)
- M A Abdallah
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
- Medical Physiology Department, Faculty of Medicine, Menoufia National University, Tukh Tanbisha, Birket ElSaba, Menoufia, Egypt
| | - Essam Omar Ibrahem
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
- Medical Physiology Department, Faculty of Medicine, Menoufia National University, Tukh Tanbisha, Birket ElSaba, Menoufia, Egypt
| | - Eman A Ali
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt.
- Clinical Pharmacology Department, Faculty of Medicine, Menoufia National University, Tukh Tanbisha, Birket ElSaba, Menoufia, Egypt.
| | - Sara G Tayel
- Anatomy and Embryology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
- Anatomy and Embryology Department, Faculty of Medicine, Menoufia National University, Tukh Tanbisha, Birket ElSaba, Menoufia, Egypt
| | - Reda A A Abo-Elsoud
- Medical Physiology Department, Faculty of Medicine, Menoufia University, Shebin El Kom, Menoufia, Egypt
- Medical Physiology Department, Faculty of Medicine, Menoufia National University, Tukh Tanbisha, Birket ElSaba, Menoufia, Egypt
| |
Collapse
|
2
|
Li W, Zhang H, Chen L, Huang C, Jiang Z, Zhou H, Zhu X, Liu X, Zheng Z, Yu Q, He Y, Gao Y, Ma J, Yang L. Cell membrane-derived nanovesicles as extracellular vesicle-mimetics in wound healing. Mater Today Bio 2025; 31:101595. [PMID: 40104636 PMCID: PMC11914519 DOI: 10.1016/j.mtbio.2025.101595] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/28/2025] [Accepted: 02/17/2025] [Indexed: 03/20/2025] Open
Abstract
Cell membrane-derived nanovesicles (NVs) have emerged as promising alternatives to extracellular vesicles (EVs) for wound healing applications, addressing the limitations of traditional EVs, which include insufficient targeting capability, low production yield, and limited drug-loading capacity. Through mechanical cell extrusion methods, NVs exhibit superior characteristics, demonstrating enhanced yield, stability, and purity compared to natural EVs. These NVs can be derived from various membrane sources, including single cell types (stem cells, blood cells, immune cells, and bacterial membranes), hybrid cell membranes and cell membranes mixed with liposomes, with each offering unique therapeutic properties. The integration of genetic engineering and surface modifications has further enhanced NV functionality, enabling precise targeting and improved drug delivery capabilities. Recent advances in NV-based therapies have demonstrated their potential across multiple biomedical applications. Although challenges persist in terms of standardization, storage stability, and clinical translation, the combination of natural cell-derived functions with artificial modification potential positions NVs as a promising platform for next-generation therapeutic delivery systems, thereby offering new possibilities in wound healing applications. Finally, we explore the challenges and future prospects of translating NV-based therapeutics into clinical practice, providing insights into the future development of this innovative approach in wound healing and tissue repair.
Collapse
Affiliation(s)
- Wenwen Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Huihui Zhang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lianglong Chen
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaoyang Huang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Ziwei Jiang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Hai Zhou
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xinxi Zhu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoyang Liu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Zesen Zheng
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Qiuyi Yu
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yufang He
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Yanbin Gao
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Jun Ma
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Lei Yang
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| |
Collapse
|
3
|
Chavda VP, Bezbaruah R, Ahmed N, Alom S, Bhattacharjee B, Nalla LV, Rynjah D, Gadanec LK, Apostolopoulos V. Proinflammatory Cytokines in Chronic Respiratory Diseases and Their Management. Cells 2025; 14:400. [PMID: 40136649 PMCID: PMC11941495 DOI: 10.3390/cells14060400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/04/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Pulmonary homeostasis can be agitated either by external environmental insults or endogenous factors produced during respiratory/pulmonary diseases. The lungs counter these insults by initiating mechanisms of inflammation as a localized, non-specific first-line defense response. Cytokines are small signaling glycoprotein molecules that control the immune response. They are formed by numerous categories of cell types and induce the movement, growth, differentiation, and death of cells. During respiratory diseases, multiple proinflammatory cytokines play a crucial role in orchestrating chronic inflammation and structural changes in the respiratory tract by recruiting inflammatory cells and maintaining the release of growth factors to maintain inflammation. The issue aggravates when the inflammatory response is exaggerated and/or cytokine production becomes dysregulated. In such instances, unresolving and chronic inflammatory reactions and cytokine production accelerate airway remodeling and maladaptive outcomes. Pro-inflammatory cytokines generate these deleterious consequences through interactions with receptors, which in turn initiate a signal in the cell, triggering a response. The cytokine profile and inflammatory cascade seen in different pulmonary diseases vary and have become fundamental targets for advancement in new therapeutic strategies for lung diseases. There are considerable therapeutic approaches that target cytokine-mediated inflammation in pulmonary diseases; however, blocking specific cytokines may not contribute to clinical benefit. Alternatively, broad-spectrum anti-inflammatory approaches are more likely to be clinically effective. Herein, this comprehensive review of the literature identifies various cytokines (e.g., interleukins, chemokines, and growth factors) involved in pulmonary inflammation and the pathogenesis of respiratory diseases (e.g., asthma, chronic obstructive pulmonary, lung cancer, pneumonia, and pulmonary fibrosis) and investigates targeted therapeutic treatment approaches.
Collapse
Affiliation(s)
- Vivek P. Chavda
- Department of Pharmaceutics and Pharmaceutical Technology, L.M. College of Pharmacy, Ahmedabad 380009, Gujarat, India
| | - Rajashri Bezbaruah
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
- Institute of Pharmacy, Assam Medical College and Hospital, Dibrugarh 786002, Assam, India
| | - Nasima Ahmed
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
| | - Shahnaz Alom
- Department of Pharmaceutical Sciences, Faculty of Science and Engineering, Dibrugarh University, Dibrugarh 786004, Assam, India; (R.B.); (N.A.); (S.A.)
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Bedanta Bhattacharjee
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Lakshmi Vineela Nalla
- Department of Pharmacology, GITAM School of Pharmacy, GITAM (Deemed to be University), Rushikonda, Visakhapatnam 530045, Andhra Pradesh, India;
| | - Damanbhalang Rynjah
- Girijananda Chowdhury Institute of Pharmaceutical Science-Tezpur, Sonitpur 784501, Assam, India; (B.B.); (D.R.)
| | - Laura Kate Gadanec
- Institute for Health and Sport, Immunology and Translational Research Group, Victoria University, Werribee, VIC 3030, Australia;
| | - Vasso Apostolopoulos
- School of Health and Biomedical Sciences, RMIT University, Melbourne, VIC 3083, Australia;
| |
Collapse
|
4
|
Jo H, Kim M, Jeoung J, Kim W, Park YH, Jung HS, Lee W, Jeoung D. Rocaglamide Suppresses Allergic Reactions by Regulating IL-4 Receptor Signaling. Molecules 2025; 30:840. [PMID: 40005151 PMCID: PMC11858170 DOI: 10.3390/molecules30040840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Rocaglamide (Roc-A), a natural phytochemical isolated from Aglaia species, is known to exert anticancer effects. Allergic inflammation can enhance the tumorigenic potential of cancer cells. We hypothesized that Roc-A could regulate allergic inflammation. Roc-A prevented an antigen from increasing the hallmarks of allergic reactions in vitro. Roc-A suppressed passive cutaneous anaphylaxis (PCA) and passive systemic anaphylaxis (PSA). RNA sequencing analysis showed that Roc-A prevented the antigen from increasing the expression of IL-4 in RBL2H3 cells. Roc-A also prevented the antigen from increasing the expression of interleukin-4 receptor (IL-4R). Roc-A was found to form a hydrogen-bonding network with residues N92 and L64 of IL-4R in a molecular docking simulation. Roc-A prevented the antigen from inducing the binding of IL-4R to JAK1. Chromatin immunoprecipitation (ChIP) assays showed that C-Jun could bind to promoter sequences of IL-4 and IL-4R. Mouse recombinant IL-4 protein increased β-hexosaminidase activity, IL-4R expression, and the hallmarks of allergic inflammation in the antigen-independent manner. Mouse recombinant IL-4 protein increased the expressions of CD163 and arghinase-1 and markers of M2 macrophages, but decreased the expression of iNOS, a marker of M1 macrophages in lung macrophages. Roc-A regulated the effects of a culture medium of antigen-stimulated RBL2H3 cells on the expressions of iNOS and arginase-1 in RAW264.7 macrophages. The blocking of IL-4 or downregulation of IL-4R exerted negative effects on the hallmarks of allergic reactions in vitro. The blocking of IL-4 or downregulation of IL-4R also exerted negative effects on PCA, and the downregulation of IL-4R exerted negative effects on PSA. An miR-34a mimic exerted negative effects on allergic reactions in vitro. The downregulation of IL-4R prevented the antigen from decreasing the expression of miR-34a in RBL2H3 cells. We identified chemicals that could bind to IL-4R via molecular docking analysis. The IL-4R docking chemical 1536801 prevented the antigen from increasing β-hexosaminidase activity and the hallmarks of allergic reactions. The IL-4R docking chemical 1536801 also exerted a negative effect on PCA. TargetScan analysis predicted miR-34a as a negative regulator of IL-4R. We found that the anti-allergic effect of Roc-A and its mechanisms were associated with miR-34a. Taken together, our results show that understanding IL-4R-mediated allergic reactions can provide clues for the development of anti-allergy therapeutics.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Dooil Jeoung
- Department of Biochemistry, Kangwon National University, Chuncheon 24341, Republic of Korea; (H.J.); (M.K.); (J.J.); (W.K.); (H.S.J.); (W.L.)
| |
Collapse
|
5
|
Banzato R, Pinheiro-Menegasso NM, Novelli FPRS, Olivo CR, Taguchi L, de Oliveira Santos S, Fukuzaki S, Teodoro WPR, Lopes FDTQS, Tibério IFLC, de Toledo-Arruda AC, Prado MAM, Prado VF, Prado CM. Alpha-7 Nicotinic Receptor Agonist Protects Mice Against Pulmonary Emphysema Induced by Elastase. Inflammation 2024; 47:958-974. [PMID: 38227123 DOI: 10.1007/s10753-023-01953-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/14/2023] [Accepted: 12/15/2023] [Indexed: 01/17/2024]
Abstract
Pulmonary emphysema is a primary component of chronic obstructive pulmonary disease (COPD), a life-threatening disorder characterized by lung inflammation and restricted airflow, primarily resulting from the destruction of small airways and alveolar walls. Cumulative evidence suggests that nicotinic receptors, especially the α7 subtype (α7nAChR), is required for anti-inflammatory cholinergic responses. We postulated that the stimulation of α7nAChR could offer therapeutic benefits in the context of pulmonary emphysema. To investigate this, we assessed the potential protective effects of PNU-282987, a selective α7nAChR agonist, using an experimental emphysema model. Male mice (C57BL/6) were submitted to a nasal instillation of porcine pancreatic elastase (PPE) (50 µl, 0.667 IU) to induce emphysema. Treatment with PNU-282987 (2.0 mg/kg, ip) was performed pre and post-emphysema induction by measuring anti-inflammatory effects (inflammatory cells, cytokines) as well as anti-remodeling and anti-oxidant effects. Elastase-induced emphysema led to an increase in the number of α7nAChR-positive cells in the lungs. Notably, both groups treated with PNU-282987 (prior to and following emphysema induction) exhibited a significant decrease in the number of α7nAChR-positive cells. Furthermore, both groups treated with PNU-282987 demonstrated decreased levels of macrophages, IL-6, IL-1β, collagen, and elastic fiber deposition. Additionally, both groups exhibited reduced STAT3 phosphorylation and lower levels of SOCS3. Of particular note, in the post-treated group, PNU-282987 successfully attenuated alveolar enlargement, decreased IL-17 and TNF-α levels, and reduced the recruitment of polymorphonuclear cells to the lung parenchyma. Significantly, it is worth noting that MLA, an antagonist of α7nAChR, counteracted the protective effects of PNU-282987 in relation to certain crucial inflammatory parameters. In summary, these findings unequivocally demonstrate the protective abilities of α7nAChR against elastase-induced emphysema, strongly supporting α7nAChR as a pivotal therapeutic target for ameliorating pulmonary emphysema.
Collapse
Affiliation(s)
- Rosana Banzato
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Nathalia M Pinheiro-Menegasso
- Department of Biosciences, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Rua Silva Jardim 136 sala 312, Santos, SP, 11015-020, Brazil
| | | | - Clarice R Olivo
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Laura Taguchi
- Department of Biosciences, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Rua Silva Jardim 136 sala 312, Santos, SP, 11015-020, Brazil
| | - Stheffany de Oliveira Santos
- Department of Biosciences, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Rua Silva Jardim 136 sala 312, Santos, SP, 11015-020, Brazil
| | - Silvia Fukuzaki
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Walcy Paganelli Rosolia Teodoro
- Rheumatology Division of the Hospital das Clínicas, Faculdade de Medicina, Universidade de São Paulo, FMUSP, São Paulo, Brazil
| | - Fernanda D T Q S Lopes
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | - Iolanda F L C Tibério
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil
| | | | - Marco Antônio M Prado
- Department of Physiology & Pharmacology, University of Western Ontario, London, Canada
- Department of Anatomy & Cell Biology, University of Western Ontario, London, Canada
| | - Vânia F Prado
- Department of Physiology & Pharmacology, University of Western Ontario, London, Canada
- Department of Anatomy & Cell Biology, University of Western Ontario, London, Canada
| | - Carla M Prado
- Department of Internal Medicine, School of Medicine, Universidade de São Paulo, São Paulo, Brazil.
- Department of Biosciences, Instituto de Saúde e Sociedade, Universidade Federal de São Paulo, Rua Silva Jardim 136 sala 312, Santos, SP, 11015-020, Brazil.
| |
Collapse
|
6
|
Lira GVDAG, da Silva GAP, Bezerra PGDM, Sarinho ESC. Avoidance of Inhaled Pollutants and Irritants in Asthma from a Salutogenic Perspective. J Asthma Allergy 2024; 17:237-250. [PMID: 38524100 PMCID: PMC10960548 DOI: 10.2147/jaa.s445864] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Accepted: 12/19/2023] [Indexed: 03/26/2024] Open
Abstract
Much is known about the role of aeroallergens in asthma, but little is described about the damage caused by inhaled pollutants and irritants to the respiratory epithelium. In this context, the most frequent pollutants and irritants inhaled in the home environment were identified, describing the possible repercussions that may occur in the respiratory tract of the pediatric population with asthma and highlighting the role of the caregiver in environmental control through a salutogenic perspective. Searches were carried out in the MEDLINE/PubMed, Web of Science, Lilacs and Scopus databases for articles considered relevant for the theoretical foundation of this integrative review, in which interactions between exposure to pollutants and inhaled irritants and lung involvement. Articles published in the last 10 years that used the following descriptors were considered: air pollution; tobacco; particulate matter; disinfectants; hydrocarbons, fluorinated; odorants; chloramines; pesticide; asthma; and beyond Antonovsky's sense of coherence. Exposure to smoke and some substances found in cleaning products, such as benzalkonium chloride, ethylenediaminetetraacetic acid and monoethanolamine, offer potential risks for sensitization and exacerbation of asthma. The vast majority of the seven main inhaled products investigated provoke irritative inflammatory reactions and oxidative imbalance in the respiratory epithelium. In turn, the caregiver's role is essential in health promotion and the clinical control of paediatric asthma. From a salutogenic point of view, pollutants and irritants inhaled at home should be carefully investigated in the clinical history so that strategies to remove or reduce exposures can be used by caregivers of children and adolescents with asthma.
Collapse
Affiliation(s)
- Georgia Véras de Araújo Gueiros Lira
- Allergy and Immunology Research Centre, Federal University of Pernambuco, Recife, PE, Brazil
- Department of Paediatrics, Federal University of Pernambuco, Recife, PE, Brazil
| | | | | | - Emanuel S C Sarinho
- Allergy and Immunology Research Centre, Federal University of Pernambuco, Recife, PE, Brazil
- Department of Paediatrics, Federal University of Pernambuco, Recife, PE, Brazil
| |
Collapse
|
7
|
Wang J, Tian F, Cao L, Du R, Tong J, Ding X, Yuan Y, Wang C. Macrophage polarization in spinal cord injury repair and the possible role of microRNAs: A review. Heliyon 2023; 9:e22914. [PMID: 38125535 PMCID: PMC10731087 DOI: 10.1016/j.heliyon.2023.e22914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 11/17/2023] [Accepted: 11/22/2023] [Indexed: 12/23/2023] Open
Abstract
The prevention, treatment, and rehabilitation of spinal cord injury (SCI) have always posed significant medical challenges. After mechanical injury, disturbances in microcirculation, edema formation, and the generation of free radicals lead to additional damage, impeding effective repair processes and potentially exacerbating further dysfunction. In this context, inflammatory responses, especially the activation of macrophages, play a pivotal role. Different phenotypes of macrophages have distinct effects on inflammation. Activation of classical macrophage cells (M1) promotes inflammation, while activation of alternative macrophage cells (M2) inhibits inflammation. The polarization of macrophages is crucial for disease healing. A non-coding RNA, known as microRNA (miRNA), governs the polarization of macrophages, thereby reducing inflammation following SCI and facilitating functional recovery. This study elucidates the inflammatory response to SCI, focusing on the infiltration of immune cells, specifically macrophages. It examines their phenotype and provides an explanation of their polarization mechanisms. Finally, this paper introduces several well-known miRNAs that contribute to macrophage polarization following SCI, including miR-155, miR-130a, and miR-27 for M1 polarization, as well as miR-22, miR-146a, miR-21, miR-124, miR-223, miR-93, miR-132, and miR-34a for M2 polarization. The emphasis is placed on their potential therapeutic role in SCI by modulating macrophage polarization, as well as the present developments and obstacles of miRNA clinical therapy.
Collapse
Affiliation(s)
- Jiawei Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Feng Tian
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Lili Cao
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Ruochen Du
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Jiahui Tong
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| | - Xueting Ding
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Yitong Yuan
- Experimental Animal Center, Shanxi Medical University, Shanxi Taiyuan, China
| | - Chunfang Wang
- School and Hospital of Stomatology, Shanxi Medical University, Shanxi Taiyuan, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Shanxi Taiyuan, China
| |
Collapse
|
8
|
Dong H, Li J, Huang X, Liu H, Gui R. Platelet-membrane camouflaged cerium nanoparticle-embedded gelatin methacryloyl hydrogel for accelerated diabetic wound healing. Int J Biol Macromol 2023; 251:126393. [PMID: 37595703 DOI: 10.1016/j.ijbiomac.2023.126393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/15/2023] [Accepted: 08/15/2023] [Indexed: 08/20/2023]
Abstract
Impaired angiogenesis and excessive inflammation are major factors contributing to delayed wound healing in diabetic patients. This study presents the development of a novel multifunctional hydrogel, Pltm@CNPs/Gel, which incorporates platelet membrane camouflaged cerium nanoparticles into a gelatin methacryloyl matrix. The Pltm@CNPs/Gel nanocomposite hydrogel was characterized and tested for its effects on platelet activation, coagulation, cell viability, anti-oxidation, and anti-inflammation in vitro. Moreover, we evaluated the wound healing potential of the hydrogel in a diabetic rat model. Our findings demonstrate that the Pltm@CNPs/Gel hydrogel possesses anti-oxidative and anti-inflammatory properties. Furthermore, it accelerates diabetic wound healing by promoting neovascularization, cell proliferation, and collagen fiber organization. This study highlights the potential of the Pltm@CNPs/Gel hydrogel as a therapeutic option for diabetic wound healing and its promising applications as a diabetic wound dressing candidate.
Collapse
Affiliation(s)
- Hang Dong
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Li
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xueyuan Huang
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Haiting Liu
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Rong Gui
- Department of Blood Transfusion, The Third Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
9
|
Jafarzadeh A, Chauhan P, Nemati M, Jafarzadeh S, Yoshimura A. Aberrant expression of suppressor of cytokine signaling (SOCS) molecules contributes to the development of allergic diseases. Clin Exp Allergy 2023; 53:1147-1161. [PMID: 37641429 DOI: 10.1111/cea.14385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 07/20/2023] [Accepted: 08/12/2023] [Indexed: 08/31/2023]
Abstract
Suppressor of cytokine signalling (SOCS) proteins bind to certain cytokine receptors, Janus kinases and signalling molecules to regulate signalling pathways, thus controlling immune and inflammatory responses. Dysregulated expression of various types of SOCS molecules was indicated in multiple types of allergic diseases. SOCS1, SOCS2, SOCS3, SOCS5, and cytokine-inducible SH2 domain protein (CISH) can differentially exert anti-allergic impacts through different mechanisms, such as suppressing Th2 cell development and activation, reducing eosinophilia, decreasing IgE production, repressing production of pro-allergic chemokines, promoting Treg cell differentiation and activation, suppressing Th17 cell differentiation and activation, increasing anti-allergic Th1 responses, inhibiting M2 macrophage polarization, modulating survival and development of mast cells, reducing pro-allergic activity of keratinocytes, and suppressing pulmonary fibrosis. Although some anti-allergic effects were attributed to SOCS3, it can perform pro-allergic impacts through several pathways, such as promoting Th2 cell development and activation, supporting eosinophilia, boosting pro-allergic activity of eosinophils, increasing IgE production, enhancing the expression of the pro-allergic chemokine receptor, reducing Treg cell differentiation, increasing pro-allergic Th9 responses, as well as supporting mucus secretion and collagen deposition. In this review, we discuss the contrasting roles of SOCS proteins in contexts of allergic disorders to provide new insights regarding the pathophysiology of these diseases and possibly explore SOCS proteins as potential therapeutic targets for alleviating allergies.
Collapse
Affiliation(s)
- Abdollah Jafarzadeh
- Department of Immunology, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Department of Immunology, School of Medicine, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Prashant Chauhan
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, České Budějovice, Czech Republic
- Faculty of Science, University of South Bohemia, České Budějovice, Czech Republic
| | - Maryam Nemati
- Department of Hematology and Laboratory Sciences, School of Para-Medicine, Kerman University of Medical Sciences, Kerman, Iran
- Immunology of Infectious Diseases Research Center, Research Institute of Basic Medical Sciences, Rafsanjan University of Medical Sciences, Rafsanjan, Iran
| | - Sara Jafarzadeh
- Student Research Committee, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Akihiko Yoshimura
- Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
10
|
Xue Z, Li Y, Xiao S, Zhang H, Xu J. FOXA2 attenuates lipopolysaccharide‑induced pneumonia by inhibiting the inflammatory response, oxidative stress and apoptosis through blocking of p38/STAT3 signaling. Exp Ther Med 2023; 26:469. [PMID: 37664675 PMCID: PMC10469380 DOI: 10.3892/etm.2023.12168] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2023] [Accepted: 07/12/2023] [Indexed: 09/05/2023] Open
Abstract
Pneumonia is a severe inflammatory disease of the lung. Forkhead box protein A2 (FOXA2) has been demonstrated to serve an important regulatory role in various pulmonary diseases; however, the role of FOXA2 in pneumonia remains to be elucidated. The present study aimed to explore the functional effects and regulatory mechanism of FOXA2 in pneumonia. An in vitro pneumonia model was induced using lipopolysaccharide (LPS) in WI-38 cells. The mRNA and protein expression levels of FOXA2 were determined by reverse transcription-quantitative PCR and western blotting, respectively. Cell viability was assessed using a Cell Counting Kit-8 assay. Inflammatory cytokines were evaluated using ELISA kits and oxidative stress markers were assessed using a malondialdehyde assay kit, superoxide dismutase assay kit and CATalase assay kit. Cell apoptosis was evaluated using flow cytometry and the caspase3 activity was determined. Western blotting was performed to examine the protein expression levels of endoplasmic reticulum stress (ERS)-associated factors. For a rescue assay, a p38 MAPK activator, U46619, was used to investigate the regulatory mechanism of FOXA2 involving p38/STAT3 signaling. FOXA2 was downregulated in LPS-induced WI-38 cells. FOXA2 overexpression alleviated LPS-induced inflammation, oxidative stress, apoptosis and ERS in WI-38 cells. Furthermore, the inhibitory effects of FOXA2 on inflammation, oxidative stress and apoptosis, as well as ERS in LPS-induced WI-38 cells were partly weakened by additional treatment with U46619. In conclusion, FOXA2 served a protective role against LPS-induced pneumonia by regulating p38/STAT3 signaling, providing a novel idea for the development of targeted therapeutic strategies for pneumonia.
Collapse
Affiliation(s)
- Zhibin Xue
- Department of Pediatrics, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Yinglin Li
- Department of Pediatrics, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Shiji Xiao
- Department of Pediatrics, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Hanqing Zhang
- Department of Pediatrics, The Affiliated Hospital of Putian University, Putian, Fujian 351100, P.R. China
| | - Jianzhang Xu
- Department of Pediatrics, Putian Children's Hospital, Putian, Fujian 351100, P.R. China
| |
Collapse
|
11
|
Jung MA, Song HK, Jo K, Lee A, Hwang YH, Ji KY, Jung DH, Cai M, Lee JY, Pyun BJ, Kim T. Gleditsia sinensis Lam. aqueous extract attenuates nasal inflammation in allergic rhinitis by inhibiting MUC5AC production through suppression of the STAT3/STAT6 pathway. Biomed Pharmacother 2023; 161:114482. [PMID: 36921533 DOI: 10.1016/j.biopha.2023.114482] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 03/05/2023] [Accepted: 03/07/2023] [Indexed: 03/16/2023] Open
Abstract
Allergic rhinitis (AR), a chronic respiratory inflammatory disease, is among the most common chronic diseases reported worldwide. Mucus hypersecretion is a critical feature of AR pathogenesis. Although the Gleditsia sinensis extract has several beneficial effects on human health, its effects on allergic inflammation have not yet been investigated. In this study, we examined the effects of G. sinensis aqueous extract (GSAE) on nasal inflammation in an ovalbumin (OVA)-induced AR mouse model. GSAE was administered orally for 1 week and then the clinical nasal symptoms were evaluated. The levels of histamine, OVA-specific immunoglobulin (Ig) E, and interleukin (IL)-13 were measured in the serum using an enzyme-linked immunosorbent assay (ELISA). Inflammatory cells were then counted in the nasal lavage fluid (NALF) and histopathology in the nasal epithelium was evaluated. STAT3/STAT6 phosphorylation was examined in primary human nasal epithelial cells (HNEpCs) using western blot analysis. Oral administration of GSAE to OVA-induced AR mice alleviated nasal clinical symptoms and reduced OVA-specific immunoglobulin E, interleukin (IL)-13, and histamine levels. The accumulation of eosinophils in nasal lavage fluid, nasal mucosa, mast cells, goblet cells, and mucin 5AC (MUC5AC) in the nasal epithelium was also inhibited by GSAE. Treatment with GSAE inhibited the production of MUC5AC in IL-4/IL-13-stimulated primary human nasal epithelial cells through the signal transducer and activator of transcription (STAT)3/STAT6 signaling pathway. These results indicated that GSAE reduces nasal inflammation suggesting that it is a potential treatment option for AR.
Collapse
Affiliation(s)
- Myung-A Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea
| | - Hyun-Kyung Song
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea
| | - Kyuhyung Jo
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea
| | - Ami Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea
| | - Youn-Hwan Hwang
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea
| | - Kon-Young Ji
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea; Center for Companion Animal New Drug Development, Jeonbuk Branch, Korea Institute of Toxicology (KIT), 30 Baekhak1-gil, Jeongeup-si 56212, the Republic of Korea
| | - Dong Ho Jung
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea
| | - Mudan Cai
- KM Science Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea
| | - Joo Young Lee
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea
| | - Bo-Jeong Pyun
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea.
| | - Taesoo Kim
- KM Convergence Research Division, Korea Institute of Oriental Medicine (KIOM), 1672 Yuseong-daero Yuseong-gu, Daejeon 34054, the Republic of Korea.
| |
Collapse
|
12
|
Zhu T, Xiao X, Dong Y, Yuan C. Neferine alleviates ovalbumin-induced asthma via MAPK signaling pathways in mice. Allergol Immunopathol (Madr) 2023; 51:135-142. [PMID: 37169571 DOI: 10.15586/aei.v51i3.840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 03/01/2023] [Indexed: 05/13/2023]
Abstract
PURPOSE To investigate the role of neferine in ovalbumin (OVA)-induced asthma, and to reveal the possible mechanism. METHODS In OVA-induced asthmatic mice, enzyme-linked-immunosorbent serologic assay was performed to evaluate the level of interleukin (IL)-4, IL-5, IL-13, immunoglobulin E (IgE) in serum and tumor necrosis factor-α (TNF-α), IL-6, IL-1β, and monocyte chemoattractant protein-1 (MCP-1) in bronchoalveolar lavage fluid (BALF). Eosinophil, neutrophil, and lymphocyte counts in BALF were calculated to assess inflammation. The pulmonary function was measured by airway resistance, peak expiratory flow (PEF) and forced expiratory volume/forced vital capacity (FEV0.4/FVC) ratio, and respiratory rate. Hematoxylin and eosin staining and Masson staining were used to evaluate lung injury. Further, Western blot analysis was conducted to detect phosphorylation of extracellular signal-regulated kinase (ERK), c-Jun N-terminal kinase (JNK), and p38 of mitogen-activated protein kinase (MAPK) signaling pathways. RESULTS Neferine, 20 mg/kg or 40 mg/kg, could significantly decrease the levels of IL-4, IL-5, IL-13, and IgE in OVA-induced serum, and that of TNF-α, IL-6, IL-1β, and MCP-1 in OVA-induced BALF. Moreover, neferine could significantly decline eosinophil, neutrophil, and lymphocyte counts in BALF. Neferine contributed to improve OVA-induced airway resistance, promoted the value of PEF and FEV0.4/FVC ratio, and recovered the respiratory rate. It also reduced mucus secretion, distribution of inflammatory and goblet cells around bronchi, and attenuated collagen deposition in lung tissues. Furthermore, neferine reduced the phosphorylation of p38, JNK, and ERK to inhibit MAPK signaling pathways. CONCLUSION Neferine relieves asthma-induced inflammatory reaction, airway resistance, and lung injury by inhibiting MAPK signaling pathways. This could serve neferine as a novel therapeutic candidate for treating asthma.
Collapse
Affiliation(s)
- Tonggang Zhu
- Department of Respiratory, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Xue Xiao
- Department of Cardiology, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Yufu Dong
- Department of Respiratory, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China
| | - Chengbo Yuan
- Department of Respiratory, Affiliated Hospital of Changchun University of Traditional Chinese Medicine, Changchun, Jilin, China;
| |
Collapse
|
13
|
Genes related to allergen exposure in allergic rhinitis: a gene-chip-based study in a mouse model. BMC Med Genomics 2022; 15:243. [PMID: 36434595 PMCID: PMC9701046 DOI: 10.1186/s12920-022-01389-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2022] [Accepted: 11/03/2022] [Indexed: 11/27/2022] Open
Abstract
BACKGROUND The typical clinical symptoms of allergic rhinitis (AR) are known to be associated with allergen exposure; however, the underlying mechanisms are not fully understood. We wanted to gain a comprehensive view of the molecular mechanisms related to allergen exposure in a well-controlled mouse model of AR. METHODS An OVA-induced AR model was developed. Two hours and 4 weeks after the last OVA challenge, AR symptoms and local immune responses were assessed. At the same time, differentially expressed genes (DEG) in nasal mucosa were identified by gene expression microarray and further analyzed by bioinformatics methods. Verification of DEG was done by quantitative RT-PCR and immunohistochemistry. RESULTS The number of nasal rubbings and sneezes, serum OVA-specific IgE concentrations, and the number of neutrophils and eosinophils in the nasal mucosa were significantly increased at 2 h and decreased at 4 weeks after the last allergen challenge compared to controls. A total of 2119 DEG were identified, and their expression dynamics were clustered into 8 profiles. Enriched functions in Profile 5, which had a similar trend to clinical features, were mainly related to inflammatory and immune response to environmental factors, eosinophils and neutrophils chemotaxis, and cell migration. Gene co-expression Network for genes from profile 5 identified BCL3, NFKB2, SOCS3, and CD53 having a higher degree. Profile 6 showed persistence of inflammatory and immune response at 4 weeks after the last allergen challenge. Olfactory and coagulation functions were enriched mainly in profiles with downward trends. CONCLUSIONS A wide range of genes with sequential cooperative action were identified to be associated with allergen exposure in AR. BCL3 may be the most vital in symptoms manifestation. Moreover, some inflammatory responses persisted for a period after allergen exposure, supporting a new treatment strategy of targeting inflammation out of season. This study may contribute to a better understanding of AR pathogenesis and provide potential therapeutic targets for AR patients.
Collapse
|
14
|
Xu Z, Ye Y, Huang G, Li Y, Guo X, Li L, Wu Y, Xu W, Nian S, Yuan Q. EphA2 recognizes Dermatophagoidespteronyssinus to mediate airway inflammation in asthma. Int Immunopharmacol 2022; 111:109106. [PMID: 35969898 DOI: 10.1016/j.intimp.2022.109106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Revised: 07/22/2022] [Accepted: 07/26/2022] [Indexed: 11/05/2022]
Abstract
Most of the asthma with low Th2 is severe steroid-resistant asthma, the exact pathogenesis of which has not yet been fully elucidated. We found that IL-6 and IL-8 were highly expressed in the sputum supernatant of severe asthma and ephrin type-A receptor 2 (EphA2) was highly expressed on bronchial epithelial cells. So, is there a connection between these two phenomena? To clarify this issue, we stimulated bronchial epithelial cells 16HBE with Dermatophagoides pteronyssinus and its compontents LPS, respectively, and detected the activation of EphA2, activation of downstream pathways and secretion of inflammatory cytokines. A mouse asthma model was established, and the therapeutic effects of inhibiting or blocking EphA2 on mouse asthma were investigated. The results showed that D. pteronyssinus and its component LPS phosphorylated EphA2 on 16HBE, activated downstream signaling pathways STAT3 and p38 MAPK, and promoted the secretion of IL-6 and IL-8. After knockout of EphA2 on 16HBE, the activation of inflammatory pathways was attenuated and the secretion of IL-6 and IL-8 was significantly reduced. Inhibition or blockade of EphA2 on mouse airways resulted in a significant reduction in airway hyperresponsiveness and airway inflammation, and a significant decrease in the expression levels of IL-6, IL-17F, IL-1α, IL-1β and TNF in bronchoalveolar lavage fluid and lung tissue. Our study uncovers a novel role for EphA2 expressed on airway epithelial cells in the pathogenesis of asthma; EphA2 recognizes D. pteronyssinus or its component LPS and promotes the secretion of IL-6 and IL-8 by airway epithelial cell, thereby mediating airway inflammation. Thus, it is possible to provide a new molecular therapy for severe asthma.
Collapse
Affiliation(s)
- Zixi Xu
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China; Medical Laboratory, Sichuan Science City Hospital, Mianyang, Sichuan, China.
| | - Yingchun Ye
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| | - Guoping Huang
- Zigong Hospital of Woman and Children Healthcare, Sichuan, China.
| | - Yi Li
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| | - Xiyuan Guo
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| | - Lin Li
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| | - Yuchuan Wu
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| | - Wenfeng Xu
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| | - Siji Nian
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| | - Qing Yuan
- Immune Mechanism and Therapy of Major Diseases of Luzhou Key Laboratory, Public Center of Experimental Technology, School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China.
| |
Collapse
|
15
|
Wang W, Xu L, Zhou L, Wan S, Jiang L. Dioscorea nipponica Makino Relieves Ovalbumin-Induced Asthma in Mice through Regulating RKIP-Mediated Raf-1/MEK/MAPK/ERK Signaling Pathway. BIOMED RESEARCH INTERNATIONAL 2022; 2022:8077058. [PMID: 35757465 PMCID: PMC9217531 DOI: 10.1155/2022/8077058] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/11/2022] [Accepted: 05/26/2022] [Indexed: 11/24/2022]
Abstract
Purpose Dioscorea nipponica Makino (DNM) is a traditional herb with multiple medicinal functions. This study is aimed at exploring the therapeutic effects of DNM on asthma and the underlying mechanisms involving RKIP-mediated MAPK signaling pathway. Methods An ovalbumin-induced asthma model was established in mice, which was further administrated with DNM and/or locostatin (RKIP inhibitor). ELISA was performed to detect the serum titers of OVA-IgE and OVA-IgG1, bronchoalveolar lavage fluid (BALF) levels of inflammation-related biomarkers, and tissue levels of oxidative stress-related biomarkers. The expression of RKIP was measured by quantitative real-time PCR, Western blot, immunohistochemistry, and immunofluorescence. HE staining was used to observe the pathological morphology of lung tissues. The protein expression of MAPK pathway-related proteins was detected by Western blot. Results Compared with the controls, the model mice exhibited significantly higher serum titers of OVA-IgE and OVA-IgG1, BALF levels of IL-6, IL-8, IL-13, TGF-β1, and MCP-1, tissue levels of MDA and ROS, lower BALF levels of IL-10 and IFN-γ, and tissue level of GSH. DNM relieved the allergic inflammatory response and oxidative stress in the model mice. DNM also recovered the downregulation of RKIP and the pathological injury of lung tissues in asthma mice. In addition, the Raf-1/MEK/MAPK/ERK pathway in the model mice was blocked by DNM. Silencing of RKIP by locostatin weakened the relieving effects of DNM on asthma through activating the Raf-1/MEK/MAPK/ERK pathway. Conclusion DNM relieves asthma via blocking the Raf-1/MEK/MAPK/ERK pathway that mediated by RKIP upregulation.
Collapse
Affiliation(s)
- Weiyi Wang
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310003, China
| | - Liying Xu
- Department of Emergency, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310003, China
| | - Lingming Zhou
- Department of Respiratory Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Shanhong Wan
- Department of Respiratory Medicine, Zhejiang Chinese Medical University, Hangzhou 310053, China
| | - Libin Jiang
- Department of Geriatric Medicine, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou 310003, China
| |
Collapse
|
16
|
Bai D, Sun T, Lu F, Shen Y, Zhang Y, Zhang B, Yu G, Li H, Hao J. Eupatilin Suppresses OVA-Induced Asthma by Inhibiting NF-κB and MAPK and Activating Nrf2 Signaling Pathways in Mice. Int J Mol Sci 2022; 23:ijms23031582. [PMID: 35163503 PMCID: PMC8836136 DOI: 10.3390/ijms23031582] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/17/2022] [Accepted: 01/24/2022] [Indexed: 12/10/2022] Open
Abstract
To investigate the effect of eupatilin in asthma treatment, we evaluated its therapeutic effect and related signal transduction in OVA-induced asthmatic mice and LPS-stimulated RAW264.7 cells. The BALF was tested for changes in lung inflammatory cells. Th2 cytokines in the BALF and OVA-IgE in the serum were measured by ELISA. H&E and PAS staining were used to evaluate histopathological changes in mouse lungs. The key proteins NF-κB, MAPK, and Nrf2 in lung tissues were quantitatively analyzed by Western blotting. Finally, we evaluated the effect of eupatilin on cytokines and related protein expression in LPS-stimulated RAW 264.7 cells in vitro. In OVA-induced asthmatic mice, eupatilin reduced the numbers of inflammatory cells, especially neutrophils and eosinophils. Eupatilin also decreased the levels of IL-5, IL-13 in the BALF and OVA-IgE in the serum. Furthermore, eupatilin inhibited the activation of NF-κB and MAPK pathways and increased the expression of Nrf2 in OVA-induced asthmatic mice. In vitro, eupatilin significantly reduced LPS-stimulated NO, IL-6, and ROS production. Additionally, the NF-κB, MAPK, and Nrf2 protein expression in LPS-stimulated RAW264.7 cells was consistent with that in OVA-induced asthmatic lung tissues. In summary, eupatilin attenuated OVA-induced asthma by regulating NF-κB, MAPK, and Nrf2 signaling pathways. These results suggest the utility of eupatilin as an anti-inflammatory drug for asthma treatment.
Collapse
Affiliation(s)
- Donghui Bai
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
| | - Tianxiao Sun
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
| | - Fang Lu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
| | - Yancheng Shen
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
| | - Yan Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
| | - Bo Zhang
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
| | - Guangli Yu
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
- Shandong Provincial Key Laboratory of Glycoscience and Glycotechnology, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
| | - Haihua Li
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
- Correspondence: (H.L.); (J.H.); Tel./Fax: +86-532-8203-1913 (J.H.)
| | - Jiejie Hao
- Key Laboratory of Marine Drugs, Ministry of Education, School of Medicine and Pharmacy, Ocean University of China, Qingdao 266003, China; (D.B.); (T.S.); (F.L.); (Y.S.); (Y.Z.); (B.Z.); (G.Y.)
- Laboratory for Marine Drugs and Bioproducts, Pilot National Laboratory for Marine Science and Technology (Qingdao), Qingdao 266237, China
- Correspondence: (H.L.); (J.H.); Tel./Fax: +86-532-8203-1913 (J.H.)
| |
Collapse
|
17
|
Zhang HY, Xie QM, Zhao CC, Sha JF, Ruan Y, Wu HM. CpG Oligodeoxynucleotides Attenuate OVA-Induced Allergic Airway Inflammation via Suppressing JNK-Mediated Endoplasmic Reticulum Stress. J Asthma Allergy 2021; 14:1399-1410. [PMID: 34848975 PMCID: PMC8619852 DOI: 10.2147/jaa.s334541] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/20/2021] [Indexed: 11/23/2022] Open
Abstract
Purpose CpG-ODN has been found to attenuate allergic airway inflammation in our previous study. Here, we aimed to further investigate whether CpG-ODN exerts such effect via regulating endoplasmic reticulum (ER) stress and revealed the underlying mechanism. Methods Five-week-old C57BL/6 mice were randomly grouped and treated with or without CpG-ODN or/and SP600125. Meantime, RAW264.7 cells were used to investigate the effect of CpG-ODN on OVA-induced ER stress in vitro. The cellularity of bronchoalveolar lavage fluid (BALF) was classified and counted after Wright-Giemsa staining. HE and PAS staining methods were applied to analyze airway inflammation. The protein levels of IL-4, IL-5, IL-13, p-JNK, JNK, CHOP, XBP1, ATF6α and GRP78 in lung tissues were detected by Western blotting. Correspondingly, the ER stress markers were detected by Western blotting and immunofluorescence in RAW264.7 cells. Results In OVA-induced allergic airway inflammation, CpG-ODN significantly suppressed inflammatory cells infiltration, goblet cell hyperplasia and the protein expression of Th2 cytokines. Moreover, OVA exposure strongly increased the activation of ER stress with higher protein expressions of CHOP, XBP1, ATF6α and GRP78. However, these OVA-induced increase of ER stress markers were markedly suppressed by CpG-ODN treatment. In addition, exposure to OVA significantly increased the phosphorylation of JNK, which was significantly reduced by CpG-ODN treatment. Remarkably, single treatment of SP600125, an antagonist of JNK, functioned similarly as CpG-ODN in mitigating allergic airway inflammation and suppressing OVA-induced activation of ER stress; however, no significant synergistic effect was evidenced by combined treatment of SP600125 and CpG-ODN. Furthermore, in OVA-stimulated RAW264.7 cells, we also found that OVA stimulation increased the expressions of ER stress markers, and CpG-ODN significantly reduced their expression levels via suppressing the phosphorylation of JNK. Conclusion These results indicated that CpG-ODN mitigates allergic airway inflammation via suppressing the activation of JNK-medicated ER stress.
Collapse
Affiliation(s)
- Hai-Yun Zhang
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Qiu-Meng Xie
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Cui-Cui Zhao
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Jia-Feng Sha
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Ya Ruan
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| | - Hui-Mei Wu
- Anhui Geriatric Institute, Department of Geriatric Respiratory and Critical Care, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, People's Republic of China.,Key Laboratory of Geriatric Molecular Medicine of Anhui Province, Hefei, Anhui, People's Republic of China.,Key Laboratory of Respiratory Disease Research and Medical Transformation of Anhui Province, Hefei, Anhui, People's Republic of China
| |
Collapse
|
18
|
Zhu Y, Sun D, Liu H, Sun L, Jie J, Luo J, Peng L, Song L. Bixin protects mice against bronchial asthma though modulating PI3K/Akt pathway. Int Immunopharmacol 2021; 101:108266. [PMID: 34678694 DOI: 10.1016/j.intimp.2021.108266] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2021] [Revised: 10/07/2021] [Accepted: 10/11/2021] [Indexed: 12/17/2022]
Abstract
Accumulating evidence has implicated the potential of natural compounds in treatment of asthma. Bixin is a natural food coloring isolated from the seeds of Bixa Orellana, which possesses anti-tumor, anti-inflammatory and antioxidative properties. Nevertheless, its therapeutic effect in asthma has not been elucidated. Our present study demonstrated that administration of Bixin suppressed allergic airway inflammation and reversed glucocorticoids resistance, as well as alleviated airway remodeling and airway hyperresponsiveness (AHR) in asthmatic mice. In vitro studies showed that Bixin treatment could inhibit the development of epithelial-mesenchymal transition (EMT) mediated by transforming growth factor beta (TGF-β) signaling. Importantly, Bixin antagonized activation of phosphatidylinositol 3-kinase/protein kinase B (PI3K/Akt) pathway both in vitro and in vivo. Above all, our findings reveal that Bixin functions as a potent antagonist of PI3K/Akt signaling to protect against allergic asthma, highlighting a novel strategy for asthma treatment based on natural products.
Collapse
Affiliation(s)
- Yingjie Zhu
- Department of Respiratory Medicine, Center For Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China
| | - Dong Sun
- Department of General Surgery, Qilu Hospital of Shandong University, Jinan 250012, Shandong, PR China
| | - Han Liu
- Department of Respiratory Medicine, Center For Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China
| | - Linzi Sun
- Department of Respiratory Medicine, Center For Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China
| | - Jing Jie
- Department of Respiratory Medicine, Center For Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China
| | - Jingjing Luo
- Department of Respiratory Medicine, Center For Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China.
| | - Liping Peng
- Department of Respiratory Medicine, Center For Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China.
| | - Lei Song
- Department of Respiratory Medicine, Center For Pathogen Biology and Infectious Diseases, The First Hospital of Jilin University, Changchun 130021, Jilin, PR China.
| |
Collapse
|
19
|
Nguyen TT. 1H/ 13C chemical shift calculations for biaryls: DFT approaches to geometry optimization. ROYAL SOCIETY OPEN SCIENCE 2021; 8:210954. [PMID: 34631126 PMCID: PMC8479412 DOI: 10.1098/rsos.210954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Accepted: 09/01/2021] [Indexed: 06/13/2023]
Abstract
Twelve common density functional methods and seven basis sets for geometry optimization were evaluated on the accuracy of 1H/13C NMR chemical shift calculations for biaryls. For these functionals, 1H shifts calculations for gas phase optimized geometries were significantly less accurate than those for in-solution optimized structures, while 13C results were not strongly influenced by geometry optimization methods and solvent effects. B3LYP, B3PW91, mPW1PW91 and ωB97XD were the best-performing functionals with lowest errors; among seven basis sets, DGDZVP2 and 6-31G(d,p) outperformed the others. The combination of these functionals and basis sets resulted in high accuracy with CMAEmin = 0.0327 ppm (0.76%) and 0.888 ppm (0.58%) for 1H and 13C, respectively. The selected functionals and basis set were validated when consistently producing optimized structures with high accuracy results for 1H and 13C chemical shift calculations of two other biaryls. This study highly recommends the IEFPCM/B3LYP, B3PW91, mPW1PW91 or ωB97XD/DGDZVP2 or 6-31G(d,p) level of theory for the geometry optimization step, especially the solvent incorporation, which would lead to high accuracy 1H/13C calculation. This work would assist in the fully structural assignments of biaryls and provide insights into in-solution biaryl conformations.
Collapse
Affiliation(s)
- Thien T Nguyen
- Institute of Research and Development, Duy Tan University, Da Nang 550000, Vietnam
- Faculty of Pharmacy, College of Medicine and Pharmacy, Duy Tan University, Da Nang 550000, Vietnam
| |
Collapse
|
20
|
Bittencourt-Mernak MI, Pinheiro NM, da Silva RC, Ponci V, Banzato R, Pinheiro AJMCR, Olivo CR, Tibério IFLC, Lima Neto LG, Santana FPR, Lago JHG, Prado CM. Effects of Eugenol and Dehydrodieugenol B from Nectandra leucantha against Lipopolysaccharide (LPS)-Induced Experimental Acute Lung Inflammation. JOURNAL OF NATURAL PRODUCTS 2021; 84:2282-2294. [PMID: 34264084 DOI: 10.1021/acs.jnatprod.1c00386] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Acute lung injury (ALI) is an important public health problem. The present work investigated whether dehydrodieugenol B treatment, a compound isolated from Brazilian plant Nectandra leucantha (Lauraceae), modulates experimental ALI and compared the observed effects to eugenol. Effects of dehydrodieugenol B in vitro in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells were evaluated. The lung and systemic inflammatory profile, lung function, and possible mechanisms involved in BALB/C male mice (6-8 weeks) with ALI induced by LPS instillation (5 mg/kg) was assayed. Dehydrodieugenol B did not affect the cell viability and inhibited the increase in NO release and IL-1β and IL-6 gene expression induced by LPS. In vivo, both compounds reduced lung edema, inflammatory cells, and the IL-6 and IL-1 β levels in bronchoalveolar lavage fluid, as well as reduced inflammatory cell infiltration and those positive to iNOS, MMP-9, and TIMP-1, and reduced the collagen content and the 8-isoprostane expression in lung tissue. Eugenol and dehydrodieugenol B also inhibited the phosphorylation of Jc-Jun-NH2 terminal Kinase (JNK), a signaling protein involved in the MAPKinase pathway. There was no effect of these compounds in lung function. Therefore, eugenol and dehydrodieugenol B ameliorates several features of experimental ALI and could be considered as a pharmacological tool to ameliorate acute lung inflammation.
Collapse
Affiliation(s)
| | - Nathalia M Pinheiro
- Department of Bioscience, Federal University of São Paulo, Santos, SP, 11015-020, Brazil
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| | - Rafael C da Silva
- Department of Biological Science, Federal University of São Paulo, Diadema, SP, 09913-030, Brazil
| | - Vitor Ponci
- Department of Biological Science, Federal University of São Paulo, Diadema, SP, 09913-030, Brazil
| | - Rosana Banzato
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| | - Aruanã J M C R Pinheiro
- Universidade CEUMA, São Luís, MA, 65075-120, Brazil
- Programa de Pós-Graduação da Rede BIONORTE, São Luís, MA, 65055-310, Brazil
| | - Clarice R Olivo
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| | - Iolanda F L C Tibério
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| | - Lídio G Lima Neto
- Universidade CEUMA, São Luís, MA, 65075-120, Brazil
- Programa de Pós-Graduação da Rede BIONORTE, São Luís, MA, 65055-310, Brazil
| | - Fernanda P R Santana
- Department of Biological Science, Federal University of São Paulo, Diadema, SP, 09913-030, Brazil
- Department of Medicine-Nephrology, Federal University of São Paulo, São Paulo, SP, 04023-062, Brazil
| | - João H G Lago
- Center of Natural Sciences and Humanities, Federal University of ABC, Santo André, SP, 09210-170, Brazil
| | - Carla M Prado
- Department of Biological Science, Federal University of São Paulo, Diadema, SP, 09913-030, Brazil
- Department of Bioscience, Federal University of São Paulo, Santos, SP, 11015-020, Brazil
- Department of Medicine, School of Medicine, University of São Paulo, São Paulo, SP, 01246-000, Brazil
| |
Collapse
|
21
|
Tirpude NV, Sharma A, Joshi R, Kumari M, Acharya V. Vitex negundo Linn. extract alleviates inflammatory aggravation and lung injury by modulating AMPK/PI3K/Akt/p38-NF-κB and TGF-β/Smad/Bcl2/caspase/LC3 cascade and macrophages activation in murine model of OVA-LPS induced allergic asthma. JOURNAL OF ETHNOPHARMACOLOGY 2021; 271:113894. [PMID: 33516930 DOI: 10.1016/j.jep.2021.113894] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Revised: 01/08/2021] [Accepted: 01/25/2021] [Indexed: 06/12/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE There is growing inclination towards developing bioactive molecule-based strategies for the management of allergic airway inflammation associated respiratory diseases. Vitex negundo Linn., also known as Nirgundi, is one such medicinal plant enriched with phytochemicals and used for inflammatory and respiratory disorders including asthma in traditional system of medicine. Preliminary studies have claimed anti-tussive and bronchodilator potential of V. negundo Linn. However, its attributes as well as molecular mechanism (s) in modulation of asthma mediated by allergic inflammation are yet to be delineated scientifically. AIM OF THE STUDY Present study attempted to assess the effectiveness of Vitex negundo leaf extract (VNLE) in mitigation of allergen induced inflammation associated asthmatic lung damage with emphasis to delineate its molecular mechanism (s). MATERIALS AND METHODS Allergic lung inflammation was established in Balb/c mice using Ovalbumin-lipopolysaccharide (OVA-LPS). Several allergic inflammatory parameters, histopathological changes, alveolar macrophage activation and signalling pathways were assessed to examine protective effects of VNLE. UHPLC-DAD-QTOF-ESI-IMS was used to characterize VLNE. RESULTS VNLE administration effectively attenuated LPS-induced oxi-inflammatory stress in macrophages suggesting its anti-inflammatory potential. Further, VNLE showed protective effect in mitigating asthmatic lung damage as evident by reversal of pathological changes including inflammatory cell influx, congestion, fibrosis, bronchial thickness and alveolar collapse observed in allergen group. VNLE suppressed expressions of inflammatory Th1/Th2 cytokines, chemokines, endopeptidases (MMPs), oxidative effector enzyme (iNOS), adhesion molecules, IL-4/IFN-γ release with simultaneous enhancement in levels of IL-10, IFN-γ, MUC3 and tight junction proteins. Subsequent mechanistic investigation revealed that OVA-LPS concomitantly enhanced phosphorylation of NF-κB, PI3K, Akt and p38MAPKs and downregulated AMPK which was categorically counteracted by VNLE treatment. VNLE also suppressed OVA-LPS induced fibrosis, apoptosis, autophagy and gap junction proteins which were affirmed by reduction in TGF-β, Smad2/3/4, Caspase9/3, Bax, LC3A/B, connexin 50, connexin 43 and enhancement in Bcl2 expression. Additionally, suppression of alveolar macrophage activation, inflammatory cells in blood and elevation of splenic CD8+T cells was demonstrated. UHPLC-DAD-QTOF-ESI-IMS revealed presence of iridoids glycoside and phenolics which might contribute these findings. CONCLUSION These findings confer protective effect of VNLE in attenuation of allergic lung inflammation and suggest that it could be considered as valuable medicinal source for developing safe natural therapeutics for mitigation of allergic inflammation during asthma.
Collapse
Affiliation(s)
- Narendra Vijay Tirpude
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India.
| | - Anamika Sharma
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Robin Joshi
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India
| | - Monika Kumari
- Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P, India
| | - Vishal Acharya
- Biotechnology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur, H.P, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, U.P. 201002, India.
| |
Collapse
|
22
|
Biseugenol Exhibited Anti-Inflammatory and Anti-Asthmatic Effects in an Asthma Mouse Model of Mixed-Granulocytic Asthma. Molecules 2020; 25:molecules25225384. [PMID: 33217892 PMCID: PMC7698799 DOI: 10.3390/molecules25225384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Revised: 09/21/2020] [Accepted: 09/24/2020] [Indexed: 11/16/2022] Open
Abstract
In the present work, the anti-inflammatory and antiasthmatic potential of biseugenol, isolated as the main component from n-hexane extract from leaves of Nectandra leucantha and chemically prepared using oxidative coupling from eugenol, was evaluated in an experimental model of mixed-granulocytic asthma. Initially, in silico studies of biseugenol showed good predictions for drug-likeness, with adherence to Lipinski’s rules of five (RO5), good Absorption, Distribution, Metabolism and Excretion (ADME) properties and no alerts for Pan-Assay Interference Compounds (PAINS), indicating adequate adherence to perform in vivo assays. Biseugenol (20 mg·kg−1) was thus administered intraperitoneally (four days of treatment) and resulted in a significant reduction in both eosinophils and neutrophils of bronchoalveolar lavage fluid in ovalbumin-sensitized mice with no statistical difference from dexamethasone (5 mg·kg−1). As for lung function parameters, biseugenol (20 mg·kg−1) significantly reduced airway and tissue damping in comparison to ovalbumin group, with similar efficacy to positive control dexamethasone. Airway hyperresponsiveness to intravenous methacholine was reduced with biseugenol but was inferior to dexamethasone in higher doses. In conclusion, biseugenol displayed antiasthmatic effects, as observed through the reduction of inflammation and airway hyperresponsiveness, with similar effects to dexamethasone, on mixed-granulocytic ovalbumin-sensitized mice.
Collapse
|